1
|
de Bejczy A, Addolorato G, Aubin HJ, Guiraud J, Korpi ER, John Nutt D, Witkiewitz K, Söderpalm B. AUD in perspective. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 175:1-19. [PMID: 38555113 DOI: 10.1016/bs.irn.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Alcohol is a major cause of pre-mature death and individual suffering worldwide, and the importance of diagnosing and treating AUD cannot be overstated. Given the global burden and the high attributable factor of alcohol in a vast number of diseases, the need for additional interventions and the development of new medicines is considered a priority by the World Health Organization (WHO). As of today, AUD is severely under-treated with a treatment gap nearing 90%, strikingly higher than that for other psychiatric disorders. Patients often seek treatment late in the progress of the disease and even among those who seek treatment only a minority receive medication, mirroring the still-prevailing stigma of the disease, and a lack of access to effective treatments, as well as a reluctance to total abstinence. To increase adherence, treatment goals should focus not only on maintaining abstinence, but also on harm reduction and psychosocial functioning. A personalised approach to AUD treatment, with a holistic view, and tailored therapy has the potential to improve AUD treatment outcomes by targeting the heterogeneity in genetics and pathophysiology, as well as reason for, and reaction to drinking. Also, the psychiatric co-morbidity rates are high in AUD and dual diagnosis can worsen symptoms and influence treatment response and should be considered in the treatment strategies.
Collapse
Affiliation(s)
- Andrea de Bejczy
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Addiction and Dependency, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Giovanni Addolorato
- Department of Medical and Surgical Sciences, Università Cattolica di Roma, Rome, Italy; Internal Medicine and Alcohol Related Disease Unit, Columbus-Gemelli Hospital, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Henri-Jean Aubin
- Université Paris-Saclay, Inserm, CESP, Villejuif, France; AP-HP, Université Paris Saclay, Villejuif, France
| | - Julien Guiraud
- Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam, The Netherlands; Vergio, Clichy, France
| | - Esa R Korpi
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - David John Nutt
- Imperial College London and GABA Labs, London, United Kingdom
| | - Katie Witkiewitz
- Department of Psychology and Center on Alcohol, Substance Use, and Addictions, University of New Mexico, Albuquerque, New Mexico, United States
| | - Bo Söderpalm
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Addiction and Dependency, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
2
|
Fanfarillo F, Ferraguti G, Lucarelli M, Fuso A, Ceccanti M, Terracina S, Micangeli G, Tarani L, Fiore M. The Impact of Alcohol-Induced Epigenetic Modifications in the Treatment of Alcohol use Disorders. Curr Med Chem 2024; 31:5837-5855. [PMID: 37828672 DOI: 10.2174/0109298673256937231004093143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/07/2023] [Accepted: 09/14/2023] [Indexed: 10/14/2023]
Abstract
Alcohol use disorders are responsible for 5.9% of all death annually and 5.1% of the global disease burden. It has been suggested that alcohol abuse can modify gene expression through epigenetic processes, namely DNA and histone methylation, histone acetylation, and microRNA expression. The alcohol influence on epigenetic mechanisms leads to molecular adaptation of a wide number of brain circuits, including the hypothalamus-hypophysis-adrenal axis, the prefrontal cortex, the mesolimbic-dopamine pathways and the endogenous opioid pathways. Epigenetic regulation represents an important level of alcohol-induced molecular adaptation in the brain. It has been demonstrated that acute and chronic alcohol exposure can induce opposite modifications in epigenetic mechanisms: acute alcohol exposure increases histone acetylation, decreases histone methylation and inhibits DNA methyltransferase activity, while chronic alcohol exposure induces hypermethylation of DNA. Some studies investigated the chromatin status during the withdrawal period and the craving period and showed that craving was associated with low methylation status, while the withdrawal period was associated with elevated activity of histone deacetylase and decreased histone acetylation. Given the effects exerted by ethanol consumption on epigenetic mechanisms, chromatin structure modifiers, such as histone deacetylase inhibitors and DNA methyltransferase inhibitors, might represent a new potential strategy to treat alcohol use disorder. Further investigations on molecular modifications induced by ethanol might be helpful to develop new therapies for alcoholism and drug addiction targeting epigenetic processes.
Collapse
Affiliation(s)
| | - Giampiero Ferraguti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Marco Lucarelli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Andrea Fuso
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Mauro Ceccanti
- SITAC, Società Italiana per il Trattamento dell'Alcolismo e le sue Complicanze, Sapienza University of Rome, Rome, Italy
| | - Sergio Terracina
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Ginevra Micangeli
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Italy
| | - Luigi Tarani
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Italy
| | - Marco Fiore
- Institute of Biochemistry and Cell Biology, IBBC-CNR, Rome, Italy
| |
Collapse
|
3
|
Mallick R, Duttaroy AK. Epigenetic modification impacting brain functions: Effects of physical activity, micronutrients, caffeine, toxins, and addictive substances. Neurochem Int 2023; 171:105627. [PMID: 37827244 DOI: 10.1016/j.neuint.2023.105627] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/06/2023] [Accepted: 10/07/2023] [Indexed: 10/14/2023]
Abstract
Changes in gene expression are involved in many brain functions. Epigenetic processes modulate gene expression by histone modification and DNA methylation or RNA-mediated processes, which is important for brain function. Consequently, epigenetic changes are also a part of brain diseases such as mental illness and addiction. Understanding the role of different factors on the brain epigenome may help us understand the function of the brain. This review discussed the effects of caffeine, lipids, addictive substances, physical activity, and pollutants on the epigenetic changes in the brain and their modulatory effects on brain function.
Collapse
Affiliation(s)
- Rahul Mallick
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Finland
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, POB 1046 Blindern, Oslo, Norway.
| |
Collapse
|
4
|
Ghare SS, Charpentier BT, Ghooray DT, Zhang J, Vadhanam MV, Reddy S, Joshi-Barve S, McClain CJ, Barve SS. Tributyrin Mitigates Ethanol-Induced Lysine Acetylation of Histone-H3 and p65-NFκB Downregulating CCL2 Expression and Consequent Liver Inflammation and Injury. Nutrients 2023; 15:4397. [PMID: 37892472 PMCID: PMC10610222 DOI: 10.3390/nu15204397] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 09/27/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
PURPOSE Chemokine-driven leukocyte infiltration and sustained inflammation contribute to alcohol-associated liver disease (ALD). Elevated hepatic CCL2 expression, seen in ALD, is associated with disease severity. However, mechanisms of CCL2 regulation are not completely elucidated. Post-translational modifications (PTMs) of proteins, particularly acetylation, modulate gene expression. This study examined the acetylation changes of promoter-associated histone-H3 and key transcription factor-NFκB in regulating hepatic CCL2 expression and subsequent inflammation and injury. Further, the effect of therapeutic modulation of the acetylation state by tributyrin (TB), a butyrate prodrug, was assessed. METHODS Hepatic CCL2 expression was assessed in mice fed control (PF) or an ethanol-containing Lieber-DeCarli (5% v/v, EF) diet for 7 weeks with or without oral administration of tributyrin (TB, 2 g/kg, 5 days/week). A chromatin immunoprecipitation (ChIP) assay evaluated promoter-associated modifications. Nuclear association between SIRT1, p300, and NFκB-p65 and acetylation changes of p65 were determined using immunoprecipitation and Western blot analyses. A Student's t-test and one-way ANOVA determined the significance. RESULTS Ethanol significantly increased promoter-associated histone-H3-lysine-9 acetylation (H3K9Ac), reflecting a transcriptionally permissive state with a resultant increase in hepatic CCL2 mRNA and protein expression. Moreover, increased lysine-310-acetylation of nuclear RelA/p65 decreased its association with SIRT1, a class III HDAC, but concomitantly increased with p300, a histone acetyltransferase. This further led to enhanced recruitment of NF-κB/p65 and RNA polymerase-II to the CCL2 promoter. Oral TB administration prevented ethanol-associated acetylation changes, thus downregulating CCL2 expression, hepatic neutrophil infiltration, and inflammation/ injury. CONCLUSION The modulation of a protein acetylation state via ethanol or TB mechanistically regulates hepatic CCL2 upregulation in ALD.
Collapse
Affiliation(s)
- Smita S. Ghare
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
- UofL Alcohol Center, University of Louisville, Louisville, KY 40202, USA
- UofL Hepatobiology COBRE, University of Louisville, Louisville, KY 40202, USA
| | - Benjamin T. Charpentier
- UofL Alcohol Center, University of Louisville, Louisville, KY 40202, USA
- Department of Anatomical Science and Neurobiology, University of Louisville, Louisville, KY 40202, USA
| | - Dushan T. Ghooray
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
- UofL Alcohol Center, University of Louisville, Louisville, KY 40202, USA
- UofL Hepatobiology COBRE, University of Louisville, Louisville, KY 40202, USA
| | - Jingwen Zhang
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
- UofL Alcohol Center, University of Louisville, Louisville, KY 40202, USA
- UofL Hepatobiology COBRE, University of Louisville, Louisville, KY 40202, USA
| | - Manicka V. Vadhanam
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
- UofL Alcohol Center, University of Louisville, Louisville, KY 40202, USA
- UofL Hepatobiology COBRE, University of Louisville, Louisville, KY 40202, USA
| | - Sreelatha Reddy
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
- UofL Alcohol Center, University of Louisville, Louisville, KY 40202, USA
- UofL Hepatobiology COBRE, University of Louisville, Louisville, KY 40202, USA
| | - Swati Joshi-Barve
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
- UofL Alcohol Center, University of Louisville, Louisville, KY 40202, USA
- UofL Hepatobiology COBRE, University of Louisville, Louisville, KY 40202, USA
| | - Craig J. McClain
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
- UofL Alcohol Center, University of Louisville, Louisville, KY 40202, USA
- UofL Hepatobiology COBRE, University of Louisville, Louisville, KY 40202, USA
- Robley Rex VA Medical Center, University of Louisville, Louisville, KY 40202, USA
| | - Shirish S. Barve
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
- UofL Alcohol Center, University of Louisville, Louisville, KY 40202, USA
- UofL Hepatobiology COBRE, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
5
|
Subramaiyam N. Insights of mitochondrial involvement in alcoholic fatty liver disease. J Cell Physiol 2023; 238:2175-2190. [PMID: 37642259 DOI: 10.1002/jcp.31100] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/31/2023] [Accepted: 08/04/2023] [Indexed: 08/31/2023]
Abstract
Alcoholic liver disease (ALD) is a global concern affecting most of the population and leading to the development of end-stage liver disease. Metabolic alterations due to increased alcohol consumption surge the hepatic accumulation of lipids and develop into a severe form of alcoholic steatohepatitis (ASH), depending on age and the consumption rate. The mitochondria in the hepatocyte actively regulate metabolic homeostasis and are disrupted in ALD pathogenesis. The increased NADH upon ethanol metabolism inhibits the mitochondrial oxidation of fatty acids, alters oxidative phosphorylation, and favors de novo lipogenesis. The higher mitochondrial respiration in early ALD increases free radical generation, whereas mitochondrial respiration is uncoupled in chronic ALD, affecting the cellular energy status. The defective glutathione importer due to excessive cholesterol loading and low adenosine triphosphate accounts for additional oxidative stress leading to hepatocyte apoptosis. The defective mitochondrial transcription machinery and sirtuins function in ALD affect mitochondrial function and biogenesis. The metabolites of ethanol metabolism epigenetically alter the gene expression profile of hepatic cell populations by modulating the promoters and sirtuins, aiding hepatic fibrosis and inflammation. The defect in mitophagy increases the accumulation of megamitochondria in hepatocytes and attracts immune cells by releasing mitochondrial damage-associated molecular patterns to initiate hepatic inflammation and ASH progression. Thus, maintaining mitochondrial lipid homeostasis and antioxidant capacity pharmacologically could provide a better outcome for ALD management.
Collapse
Affiliation(s)
- Nithyananthan Subramaiyam
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
6
|
Bhatia S, Bodenstein D, Cheng AP, Wells PG. Altered Epigenetic Marks and Gene Expression in Fetal Brain, and Postnatal Behavioural Disorders, Following Prenatal Exposure of Ogg1 Knockout Mice to Saline or Ethanol. Cells 2023; 12:2308. [PMID: 37759530 PMCID: PMC10527575 DOI: 10.3390/cells12182308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Oxoguanine glycosylase 1 (OGG1) is widely known to repair the reactive oxygen species (ROS)-initiated DNA lesion 8-oxoguanine (8-oxoG), and more recently was shown to act as an epigenetic modifier. We have previously shown that saline-exposed Ogg1 -/- knockout progeny exhibited learning and memory deficits, which were enhanced by in utero exposure to a single low dose of ethanol (EtOH) in both Ogg1 +/+ and -/- progeny, but more so in Ogg1 -/- progeny. Herein, OGG1-deficient progeny exposed in utero to a single low dose of EtOH or its saline vehicle exhibited OGG1- and/or EtOH-dependent alterations in global histone methylation and acetylation, DNA methylation and gene expression (Tet1 (Tet Methylcytosine Dioxygenase 1), Nlgn3 (Neuroligin 3), Hdac2 (Histone Deacetylase 2), Reln (Reelin) and Esr1 (Estrogen Receptor 1)) in fetal brains, and behavioural changes in open field activity, social interaction and ultrasonic vocalization, but not prepulse inhibition. OGG1- and EtOH-dependent changes in Esr1 and Esr2 mRNA and protein levels were sex-dependent, as was the association of Esr1 gene expression with gene activation mark histone H3 lysine 4 trimethylation (H3K4me3) and gene repression mark histone H3 lysine 27 trimethylation (H3K27me3) measured via ChIP-qPCR. The OGG1-dependent changes in global epigenetic marks and gene/protein expression in fetal brains, and postnatal behavioural changes, observed in both saline- and EtOH-exposed progeny, suggest the involvement of epigenetic mechanisms in developmental disorders mediated by 8-oxoG and/or OGG1. Epigenetic effects of OGG1 may be involved in ESR1-mediated gene regulation, which may be altered by physiological and EtOH-enhanced levels of ROS formation, possibly contributing to sex-dependent developmental disorders observed in Ogg1 knockout mice. The OGG1- and EtOH-dependent associations provide a basis for more comprehensive mechanistic studies to determine the causal involvement of oxidative DNA damage and epigenetic changes in ROS-mediated neurodevelopmental disorders.
Collapse
Affiliation(s)
- Shama Bhatia
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada; (S.B.); (A.P.C.)
- Centre for Pharmaceutical Oncology, Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - David Bodenstein
- Department of Pharmacology & Toxicology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Ashley P. Cheng
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada; (S.B.); (A.P.C.)
- Centre for Pharmaceutical Oncology, Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Peter G. Wells
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada; (S.B.); (A.P.C.)
- Centre for Pharmaceutical Oncology, Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
- Department of Pharmacology & Toxicology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada;
| |
Collapse
|
7
|
McGinnis CD, Jennings EQ, Harris PS, Galligan JJ, Fritz KS. Biochemical Mechanisms of Sirtuin-Directed Protein Acylation in Hepatic Pathologies of Mitochondrial Dysfunction. Cells 2022; 11:cells11132045. [PMID: 35805129 PMCID: PMC9266223 DOI: 10.3390/cells11132045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/06/2022] [Accepted: 06/10/2022] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial protein acetylation is associated with a host of diseases including cancer, Alzheimer’s, and metabolic syndrome. Deciphering the mechanisms regarding how protein acetylation contributes to disease pathologies remains difficult due to the complex diversity of pathways targeted by lysine acetylation. Specifically, protein acetylation is thought to direct feedback from metabolism, whereby nutritional status influences mitochondrial pathways including beta-oxidation, the citric acid cycle, and the electron transport chain. Acetylation provides a crucial connection between hepatic metabolism and mitochondrial function. Dysregulation of protein acetylation throughout the cell can alter mitochondrial function and is associated with numerous liver diseases, including non-alcoholic and alcoholic fatty liver disease, steatohepatitis, and hepatocellular carcinoma. This review introduces biochemical mechanisms of protein acetylation in the regulation of mitochondrial function and hepatic diseases and offers a viewpoint on the potential for targeted therapies.
Collapse
Affiliation(s)
- Courtney D. McGinnis
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (C.D.M.); (P.S.H.)
| | - Erin Q. Jennings
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA; (E.Q.J.); (J.J.G.)
| | - Peter S. Harris
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (C.D.M.); (P.S.H.)
| | - James J. Galligan
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA; (E.Q.J.); (J.J.G.)
| | - Kristofer S. Fritz
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (C.D.M.); (P.S.H.)
- Correspondence:
| |
Collapse
|
8
|
Finelli R, Mottola F, Agarwal A. Impact of Alcohol Consumption on Male Fertility Potential: A Narrative Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 19:ijerph19010328. [PMID: 35010587 PMCID: PMC8751073 DOI: 10.3390/ijerph19010328] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/11/2021] [Accepted: 12/23/2021] [Indexed: 12/13/2022]
Abstract
Alcohol abuse disorder is a serious condition, implicating more than 15 million people aged 12 years and older in 2019 in the United States. Ethanol (or ethyl alcohol) is mainly oxidized in the liver, resulting in the synthesis of acetaldehyde and acetate, which are toxic and carcinogenic metabolites, as well as in the generation of a reductive cellular environment. Moreover, ethanol can interact with lipids, generating fatty acid ethyl esters and phosphatidylethanol, which interfere with physiological cellular pathways. This narrative review summarizes the impact of excessive alcohol consumption on male fertility by describing its metabolism and how ethanol consumption may induce cellular damage. Furthermore, the impact of alcohol consumption on hormonal regulation, semen quality, and genetic and epigenetic regulations is discussed based on evidence from animal and human studies, focusing on the consequences on the offspring. Finally, the limitations of the current evidence are discussed. Our review highlights the association between chronic alcohol consumption and poor semen quality, mainly due to the development of oxidative stress, as well as its genotoxic impact on hormonal regulation and DNA integrity, affecting the offspring’s health. New landscapes of investigation are proposed for the identification of molecular markers for alcohol-associated infertility, with a focus on advanced OMICS-based approaches applied to the analysis of semen samples.
Collapse
Affiliation(s)
- Renata Finelli
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH 44106, USA;
| | - Filomena Mottola
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, 81100 Caserta, Italy;
| | - Ashok Agarwal
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH 44106, USA;
- Correspondence: ; Tel.: +1-(214)-444-9485
| |
Collapse
|
9
|
Hoes L, Dok R, Verstrepen KJ, Nuyts S. Ethanol-Induced Cell Damage Can Result in the Development of Oral Tumors. Cancers (Basel) 2021; 13:cancers13153846. [PMID: 34359747 PMCID: PMC8345464 DOI: 10.3390/cancers13153846] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Alcohol consumption is linked to 26.4% of all lip and oral cavity cancer cases worldwide. Despite this clear causal relationship, the exact molecular mechanisms by which ethanol damages cells are still under investigation. It is well-established that the metabolism of ethanol plays an important role. Ethanol metabolism yields reactive metabolites that can directly damage the DNA. If the damage is repaired incorrectly, mutations can be fixed in the DNA sequence. Whenever mutations affect key regulatory genes, for instance cell cycle regulating genes, uncontrolled cell growth can be the consequence. Recently, global patterns of mutations have been identified. These so-called mutational signatures represent a fingerprint of the different mutational processes over time. Interestingly, there were ethanol-related signatures discovered that did not associate with ethanol metabolism. This finding highlights there might be other molecular effects of ethanol that are yet to be discovered. Abstract Alcohol consumption is an underestimated risk factor for the development of precancerous lesions in the oral cavity. Although alcohol is a well-accepted recreational drug, 26.4% of all lip and oral cavity cancers worldwide are related to heavy drinking. Molecular mechanisms underlying this carcinogenic effect of ethanol are still under investigation. An important damaging effect comes from the first metabolite of ethanol, being acetaldehyde. Concentrations of acetaldehyde detected in the oral cavity are relatively high due to the metabolization of ethanol by oral microbes. Acetaldehyde can directly damage the DNA by the formation of mutagenic DNA adducts and interstrand crosslinks. Additionally, ethanol is known to affect epigenetic methylation and acetylation patterns, which are important regulators of gene expression. Ethanol-induced hypomethylation can activate the expression of oncogenes which subsequently can result in malignant transformation. The recent identification of ethanol-related mutational signatures emphasizes the role of acetaldehyde in alcohol-associated carcinogenesis. However, not all signatures associated with alcohol intake also relate to acetaldehyde. This finding highlights that there might be other effects of ethanol yet to be discovered.
Collapse
Affiliation(s)
- Lore Hoes
- Laboratory for Systems Biology, VIB-KU Leuven Center for Microbiology, 3000 Leuven, Belgium; (L.H.); (K.J.V.)
- Laboratory of Genetics and Genomics, Centre for Microbial and Plant Genetics, KU Leuven, 3000 Leuven, Belgium
- Laboratory of Experimental Radiotherapy, Department of Oncology, KU Leuven, 3000 Leuven, Belgium;
| | - Rüveyda Dok
- Laboratory of Experimental Radiotherapy, Department of Oncology, KU Leuven, 3000 Leuven, Belgium;
| | - Kevin J. Verstrepen
- Laboratory for Systems Biology, VIB-KU Leuven Center for Microbiology, 3000 Leuven, Belgium; (L.H.); (K.J.V.)
- Laboratory of Genetics and Genomics, Centre for Microbial and Plant Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Sandra Nuyts
- Laboratory of Experimental Radiotherapy, Department of Oncology, KU Leuven, 3000 Leuven, Belgium;
- Department of Radiation Oncology, Leuven Cancer Institute, University Hospital Leuven, 3000 Leuven, Belgium
- Correspondence: ; Tel.: +32-1634-7600; Fax: +32-1634-7623
| |
Collapse
|
10
|
Siomek-Gorecka A, Dlugosz A, Czarnecki D. The Molecular Basis of Alcohol Use Disorder (AUD). Genetics, Epigenetics, and Nutrition in AUD: An Amazing Triangle. Int J Mol Sci 2021; 22:ijms22084262. [PMID: 33924016 PMCID: PMC8072802 DOI: 10.3390/ijms22084262] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/20/2022] Open
Abstract
Alcohol use disorder (AUD) is a very common and complex disease, as alcohol is the most widely used addictive drug in the world. This disorder has an enormous impact on public health and social and private life, and it generates a huge number of social costs. Alcohol use stimulates hypothalamic-pituitary-adrenal (HPA) axis responses and is the cause of many physical and social problems (especially liver disease and cancer), accidental injury, and risky sexual behavior. For years, researchers have been trying to identify the genetic basis of alcohol use disorder, the molecular mechanisms responsible for its development, and an effective form of therapy. Genetic and environmental factors are known to contribute to the development of AUD, and the expression of genes is a complicated process that depends on epigenetic modulations. Dietary nutrients, such as vitamins, may serve as one these modulators, as they have a direct impact on epigenomes. In this review, we connect gathered knowledge from three emerging fields-genetics, epigenetics, and nutrition-to form an amazing triangle relating to alcohol use disorder.
Collapse
Affiliation(s)
- Agnieszka Siomek-Gorecka
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-095 Bydgoszcz, Poland
- Correspondence: ; Tel.: +48-52-585-37-48
| | - Anna Dlugosz
- Department of Engineering and Chemical and Food Analytics, Faculty of Chemical Technology and Engineering, UTP University of Science and Technology, 85-326 Bydgoszcz, Poland;
| | - Damian Czarnecki
- Department of Preventive Nursing, Faculty of Health Sciences, L. Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-821 Bydgoszcz, Poland;
| |
Collapse
|
11
|
Dulman RS, Wandling GM, Pandey SC. Epigenetic mechanisms underlying pathobiology of alcohol use disorder. CURRENT PATHOBIOLOGY REPORTS 2020; 8:61-73. [PMID: 33747641 DOI: 10.1007/s40139-020-00210-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Purpose of review Chronic alcohol use is a worldwide problem with multifaceted consequences including multiplying medical costs and sequelae, societal effects like drunk driving and assault, and lost economic productivity. These large-scale outcomes are driven by the consumption of ethanol, a small permeable molecule that has myriad effects in the human body, particularly in the liver and brain. In this review, we have summarized effects of acute and chronic alcohol consumption on epigenetic mechanisms that may drive pathobiology of Alcohol Use Disorder (AUD) while identifying areas of need for future research. Recent findings Epigenetics has emerged as an interesting field of biology at the intersection of genetics and the environment, and ethanol in particular has been identified as a potent modulator of the epigenome with various effects on DNA methylation, histone modifications, and non-coding RNAs. These changes alter chromatin dynamics and regulate gene expression that contribute to behavioral and physiological changes leading to the development of AUD psychopathology and cancer pathology. Summary Evidence and discussion presented here from preclinical results and available translational studies have increased our knowledge of the epigenetic effects of alcohol consumption. These studies have identified targets that can be used to develop better therapies to reduce chronic alcohol abuse and mitigate its societal burden and pathophysiology.
Collapse
Affiliation(s)
- Russell S Dulman
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Gabriela M Wandling
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, USA.,Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
12
|
Chen Z, Li S, Guo L, Peng X, Liu Y. Prenatal alcohol exposure induced congenital heart diseases: From bench to bedside. Birth Defects Res 2020; 113:521-534. [PMID: 32578335 DOI: 10.1002/bdr2.1743] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 05/22/2020] [Accepted: 05/23/2020] [Indexed: 12/27/2022]
Abstract
Alcohol consumption is increasing worldwide. Many child-bearing-aged women consume alcohol during pregnancy, intentionally or unintentionally, thereby increasing the potential risk for severe congenital diseases. Congenital heart disease (CHD) is the most common birth defect worldwide and can result from both hereditary and acquired factors. Prenatal alcohol exposure (PAE) is considered a key factor that leads to teratogenesis in CHD and its specific phenotypes, especially defects of the cardiac septa, cardiac valves, cardiac canals, and great arteries, adjacent to the chambers, both in animal experiments and clinical retrospective studies. The mechanisms underlying CHD and its phenotypes caused by PAE are associated with changes in retinoic acid biosynthesis and its signaling pathway, apoptosis and defective function of cardiac neural crest cells, disturbance of the Wntβ-catenin signaling pathway, suppression of bone morphogenetic protein (BMP) signaling, and other epigenetic mechanisms. Drug supplements and early diagnosis can help prevent PAE from inducing CHDs.
Collapse
Affiliation(s)
- Zhiyan Chen
- Department of Basic Medical Sciences, Sichuan Vocational College of Health and Rehabilitation, Zigong, Sichuan, China.,Department of Research, Zigong First People's Hospital, Zigong, Sichuan, China
| | - Sheng Li
- Department of Basic Medical Sciences, Sichuan Vocational College of Health and Rehabilitation, Zigong, Sichuan, China.,Department of Research, Zigong First People's Hospital, Zigong, Sichuan, China
| | - Linghong Guo
- Department of Pharmacology, West China School of Basic Sciences & Forensic Medicine; Animal Research Institute, Sichuan University, Chengdu, Sichuan, China
| | - Xu Peng
- Department of Pharmacology, West China School of Basic Sciences & Forensic Medicine; Animal Research Institute, Sichuan University, Chengdu, Sichuan, China
| | - Yin Liu
- Department of Basic Medical Sciences, Sichuan Vocational College of Health and Rehabilitation, Zigong, Sichuan, China.,Department of Research, Zigong First People's Hospital, Zigong, Sichuan, China.,Department of Pharmacology, West China School of Basic Sciences & Forensic Medicine; Animal Research Institute, Sichuan University, Chengdu, Sichuan, China.,Department of Anesthesiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
13
|
Conner KE, Bottom RT, Huffman KJ. The Impact of Paternal Alcohol Consumption on Offspring Brain and Behavioral Development. Alcohol Clin Exp Res 2019; 44:125-140. [PMID: 31746471 DOI: 10.1111/acer.14245] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Fetal alcohol spectrum disorders (FASD) describe the wide array of long-lasting developmental abnormalities in offspring due to prenatal alcohol (ethanol [EtOH]) exposure via maternal gestational drinking. Although the teratogenic consequences of prenatal EtOH exposure, are apparent, the effects of preconception paternal EtOH exposure (PatEE) are still unclear. Previous research suggests that PatEE can induce molecular changes and abnormal behavior in the offspring. However, it is not known whether PatEE impacts the development of the neocortex and behavior in offspring as demonstrated in maternal consumption models of FASD (J Neurosci, 33, 2013, 18893). METHODS In this study, we utilized a novel mouse model of PatEE where male mice self-administered 25% EtOH for an extended period prior to conception, generating indirect exposure to the offspring through the paternal germline. Following mating, we examined the effects of PatEE on offspring neocortical development at postnatal day (P) 0 and evaluated several aspects of behavior at both P20 and P30 using a battery of behavioral assays. RESULTS PatEE resulted in significant impact on neocortical development, including abnormal patterns of gene expression within the neocortex at P0 and subtle alterations in patterns of intraneocortical connections. Additionally, PatEE mice exhibited a sex-specific increase in activity and sensorimotor integration deficits at P20, and decreased balance, coordination, and short-term motor learning at P30. This suggests that PatEE may generate long-lasting, sex-specific effects on offspring behavior. CONCLUSIONS These results demonstrate that the developmental impact of preconception PatEE is more harmful than previously thought and provide additional insights into the biological mechanisms that may underlie atypical behavior observed in children of alcoholic fathers.
Collapse
Affiliation(s)
- Kathleen E Conner
- Interdepartmental Neuroscience Program, University of California, Riverside, Riverside, California
| | - Riley T Bottom
- Interdepartmental Neuroscience Program, University of California, Riverside, Riverside, California
| | - Kelly J Huffman
- Interdepartmental Neuroscience Program, University of California, Riverside, Riverside, California.,Department of Psychology, University of California, Riverside, Riverside, California
| |
Collapse
|
14
|
Persistence of Burkholderia thailandensis E264 in lung tissue after a single binge alcohol episode. PLoS One 2019; 14:e0218147. [PMID: 31821337 PMCID: PMC6903738 DOI: 10.1371/journal.pone.0218147] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023] Open
Abstract
Background Binge drinking, an increasingly common form of alcohol use disorder, is associated with substantial morbidity and mortality; yet, its effects on the immune system’s ability to defend against infectious agents are poorly understood. Burkholderia pseudomallei, the causative agent of melioidosis can occur in healthy humans, yet binge alcohol intoxication is increasingly being recognized as a major risk factor. Although our previous studies demonstrated that binge alcohol exposure increased B. pseudomallei near-neighbor virulence in vivo and increased paracellular diffusion and intracellular invasion, no experimental studies have examined the extent to which bacterial and alcohol dosage play a role in disease progression. In addition, the temporal effects of a single binge alcohol dose prior to infection has not been examined in vivo. Principal findings In this study, we used B. thailandensis E264 a close genetic relative of B. pseudomallei, as useful BSL-2 model system. Eight-week-old female C57BL/6 mice were utilized in three distinct animal models to address the effects of 1) bacterial dosage, 2) alcohol dosage, and 3) the temporal effects, of a single binge alcohol episode. Alcohol was administered comparable to human binge drinking (≤ 4.4 g/kg) or PBS intraperitoneally before a non-lethal intranasal infection. Bacterial colonization of lung and spleen was increased in mice administered alcohol even after bacterial dose was decreased 10-fold. Lung and not spleen tissue were colonized even after alcohol dosage was decreased 20 times below the U.S legal limit. Temporally, a single binge alcohol episode affected lung bacterial colonization for more than 24 h after alcohol was no longer detected in the blood. Pulmonary and splenic cytokine expression (TNF-α, GM-CSF) remained suppressed, while IL-12/p40 increased in mice administered alcohol 6 or 24 h prior to infection. Increased lung and not intestinal bacterial invasion was observed in human and murine non-phagocytic epithelial cells exposed to 0.2% v/v alcohol in vitro. Conclusions Our results indicate that the effects of a single binge alcohol episode are tissue specific. A single binge alcohol intoxication event increases bacterial colonization in mouse lung tissue even after very low BACs and decreases the dose required to colonize the lungs with less virulent B. thailandensis. Additionally, the temporal effects of binge alcohol alters lung and spleen cytokine expression for at least 24 h after alcohol is detected in the blood. Delayed recovery in lung and not spleen tissue may provide a means for B. pseudomallei and near-neighbors to successfully colonize lung tissue through increased intracellular invasion of non-phagocytic cells in patients with hazardous alcohol intake.
Collapse
|
15
|
Shukla SD, Restrepo R, Aroor AR, Liu X, Lim RW, Franke JD, Ford DA, Korthuis RJ. Binge Alcohol Is More Injurious to Liver in Female than in Male Rats: Histopathological, Pharmacologic, and Epigenetic Profiles. J Pharmacol Exp Ther 2019; 370:390-398. [PMID: 31262967 PMCID: PMC6695503 DOI: 10.1124/jpet.119.258871] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 06/19/2019] [Indexed: 12/20/2022] Open
Abstract
Binge alcohol consumption is a health problem, but differences between the sexes remain poorly defined. We have examined the in vivo effects of three acute, repeat binge alcohol administration on the liver in male and female rats. Sprague-Dawley rats were gavaged with alcohol (5 g/kg body weight) three times at 12-hour intervals. Blood and liver tissues were collected 4 hours after the last binge ethanol. Subsequently, several variables were analyzed. Compared with male rats, females had higher levels of blood alcohol, alanine aminotransferase, and triglycerides. Liver histology showed increased lipid vesicles that were larger in females. Protein levels of liver cytochrome P4502E1 were higher in the liver of females than in the liver of males after binge. Hepatic phospho-extracellular signal-regulated kinase 1/2 and phosph-p38 mitogen-activated protein kinase levels were lower in females compared with males after binge alcohol, but no differences were found in the phospho-C-jun N-terminal kinase levels. Peroxisome proliferator-activated receptor γ-coactivator 1α and cyclic AMP response element binding (CREB) protein levels increased more in female than in male livers; however, increases in phospho-CREB levels were lower in females. Remarkably, c-fos was reduced substantially in the livers of females, but no differences in c-myc protein were found. Binge ethanol caused elevation in acetylated (H3AcK9) and phosphoacetylated (H3AcK9PS10) histone H3 in both sexes but without any difference. Binge alcohol caused differential alterations in the levels of various species of phosphatidylethanol and a larger increase in the diacylglycerol kinase-α protein levels in the liver of female rats compared with male rats. These data demonstrate, for the first time, similarities and differences in the sex-specific responses to repeat binge alcohol leading to an increased susceptibility of female rats to have liver injury in vivo. SIGNIFICANCE STATEMENT: This study examines the molecular responses of male and female rat livers to acute binge alcohol in vivo and demonstrates significant differences in the susceptibility between sexes.
Collapse
Affiliation(s)
- Shivendra D Shukla
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia (S.D.S.,R.R.,A.R.A.,X.L.,R.W.L.,R.J.K.), and Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University, St. Louis (J.D.F.,D.A.F.), Missouri
| | - Ricardo Restrepo
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia (S.D.S.,R.R.,A.R.A.,X.L.,R.W.L.,R.J.K.), and Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University, St. Louis (J.D.F.,D.A.F.), Missouri
| | - Annayya R Aroor
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia (S.D.S.,R.R.,A.R.A.,X.L.,R.W.L.,R.J.K.), and Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University, St. Louis (J.D.F.,D.A.F.), Missouri
| | - Xuanyou Liu
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia (S.D.S.,R.R.,A.R.A.,X.L.,R.W.L.,R.J.K.), and Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University, St. Louis (J.D.F.,D.A.F.), Missouri
| | - Robert W Lim
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia (S.D.S.,R.R.,A.R.A.,X.L.,R.W.L.,R.J.K.), and Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University, St. Louis (J.D.F.,D.A.F.), Missouri
| | - Jacob D Franke
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia (S.D.S.,R.R.,A.R.A.,X.L.,R.W.L.,R.J.K.), and Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University, St. Louis (J.D.F.,D.A.F.), Missouri
| | - David A Ford
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia (S.D.S.,R.R.,A.R.A.,X.L.,R.W.L.,R.J.K.), and Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University, St. Louis (J.D.F.,D.A.F.), Missouri
| | - Ronald J Korthuis
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia (S.D.S.,R.R.,A.R.A.,X.L.,R.W.L.,R.J.K.), and Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University, St. Louis (J.D.F.,D.A.F.), Missouri
| |
Collapse
|
16
|
Petrelli B, Bendelac L, Hicks GG, Fainsod A. Insights into retinoic acid deficiency and the induction of craniofacial malformations and microcephaly in fetal alcohol spectrum disorder. Genesis 2019; 57:e23278. [DOI: 10.1002/dvg.23278] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Berardino Petrelli
- Regenerative Medicine Program and the Department of Biochemistry & Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health SciencesUniversity of Manitoba Winnipeg Manitoba Canada
| | - Liat Bendelac
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel‐CanadaFaculty of Medicine, Hebrew University Jerusalem Israel
| | - Geoffrey G. Hicks
- Regenerative Medicine Program and the Department of Biochemistry & Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health SciencesUniversity of Manitoba Winnipeg Manitoba Canada
| | - Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel‐CanadaFaculty of Medicine, Hebrew University Jerusalem Israel
| |
Collapse
|
17
|
Molina PE, Simon L, Amedee AM, Welsh DA, Ferguson TF. Impact of Alcohol on HIV Disease Pathogenesis, Comorbidities and Aging: Integrating Preclinical and Clinical Findings. Alcohol Alcohol 2018; 53:439-447. [PMID: 29546271 DOI: 10.1093/alcalc/agy016] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 03/01/2018] [Indexed: 12/12/2022] Open
Abstract
Short Summary : Effective combined antiretroviral therapy regimens have extended survival of persons living with HIV (PLWH). Heavy alcohol consumption is common in PLWH. This overview integrates evidence from clinical and preclinical research to identify salient alcohol-related mechanisms and comorbidities contributing to disease pathogenesis and accelerated aging and senescence in PLWH.
Collapse
Affiliation(s)
- Patricia E Molina
- Comprehensive Alcohol-HIV/AIDS Research Center and Alcohol and Drug Abuse Center of Excellence, LSUHSC, 1901 Perdido St., New Orleans, LA, USA
| | - Liz Simon
- Comprehensive Alcohol-HIV/AIDS Research Center and Alcohol and Drug Abuse Center of Excellence, LSUHSC, 1901 Perdido St., New Orleans, LA, USA
| | - Angela M Amedee
- Comprehensive Alcohol-HIV/AIDS Research Center and Alcohol and Drug Abuse Center of Excellence, LSUHSC, 1901 Perdido St., New Orleans, LA, USA
| | - David A Welsh
- Comprehensive Alcohol-HIV/AIDS Research Center and Alcohol and Drug Abuse Center of Excellence, LSUHSC, 1901 Perdido St., New Orleans, LA, USA
| | - Tekeda F Ferguson
- Comprehensive Alcohol-HIV/AIDS Research Center and Alcohol and Drug Abuse Center of Excellence, LSUHSC, 1901 Perdido St., New Orleans, LA, USA
| |
Collapse
|
18
|
Shabtai Y, Fainsod A. Competition between ethanol clearance and retinoic acid biosynthesis in the induction of fetal alcohol syndrome. Biochem Cell Biol 2018; 96:148-160. [DOI: 10.1139/bcb-2017-0132] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Several models have been proposed to explain the neurodevelopmental syndrome induced by exposure of human embryos to alcohol, which is known as fetal alcohol spectrum disorder (FASD). One of the proposed models suggests a competition for the enzymes required for the biosynthesis of retinoic acid. The outcome of such competition is development under conditions of reduced retinoic acid signaling. Retinoic acid is one of the biologically active metabolites of vitamin A (retinol), and regulates numerous embryonic and differentiation processes. The developmental malformations characteristic of FASD resemble those observed in vitamin A deficiency syndrome as well as from inhibition of retinoic acid biosynthesis or signaling in experimental models. There is extensive biochemical and enzymatic overlap between ethanol clearance and retinoic acid biosynthesis. Several lines of evidence suggest that in the embryo, the competition takes place between acetaldehyde and retinaldehyde for the aldehyde dehydrogenase activity available. In adults, this competition also extends to the alcohol dehydrogenase activity. Ethanol-induced developmental defects can be ameliorated by increasing the levels of retinol, retinaldehyde, or retinaldehyde dehydrogenase. Acetaldehyde inhibits the production of retinoic acid by retinaldehyde dehydrogenase, further supporting the competition model. All of the evidence supports the reduction of retinoic acid signaling as the etiological trigger in the induction of FASD.
Collapse
Affiliation(s)
- Yehuda Shabtai
- Department of Cellular Biochemistry and Cancer Research, Institute for Medical Research Israel–Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
- Department of Cellular Biochemistry and Cancer Research, Institute for Medical Research Israel–Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Abraham Fainsod
- Department of Cellular Biochemistry and Cancer Research, Institute for Medical Research Israel–Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
- Department of Cellular Biochemistry and Cancer Research, Institute for Medical Research Israel–Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| |
Collapse
|
19
|
Abstract
The widespread and rapidly increasing trend of binge drinking is accompanied by a concomitant rise in the prevalence of trauma patients under the influence of alcohol at the time of their injury. Epidemiological evidence suggests up to half of all adult burn patients are intoxicated at the time of admission, and the presence of alcohol is an independent risk factor for death in the early stages post burn. As the major site of alcohol metabolism and toxicity, the liver is a critical determinant of postburn outcome, and experimental evidence implies an injury threshold exists beyond which burn-induced hepatic derangement is observed. Alcohol may lower this threshold for postburn hepatic damage through a variety of mechanisms including modulation of extrahepatic events, alteration of the gut-liver axis, and changes in signaling pathways. The direct and indirect effects of alcohol may prime the liver for the second-hit of many overlapping physiologic responses to burn injury. In an effort to gain a deeper understanding of how alcohol potentiates postburn hepatic damage, the authors summarize possible mechanisms by which alcohol modulates the postburn hepatic response.
Collapse
|
20
|
Ghosh Dastidar S, Warner JB, Warner DR, McClain CJ, Kirpich IA. Rodent Models of Alcoholic Liver Disease: Role of Binge Ethanol Administration. Biomolecules 2018; 8:biom8010003. [PMID: 29342874 PMCID: PMC5871972 DOI: 10.3390/biom8010003] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 12/14/2022] Open
Abstract
Both chronic and acute (binge) alcohol drinking are important health and economic concerns worldwide and prominent risk factors for the development of alcoholic liver disease (ALD). There are no FDA-approved medications to prevent or to treat any stage of ALD. Therefore, discovery of novel therapeutic strategies remains a critical need for patients with ALD. Relevant experimental animal models that simulate human drinking patterns and mimic the spectrum and severity of alcohol-induced liver pathology in humans are critical to our ability to identify new mechanisms and therapeutic targets. There are several animal models currently in use, including the most widely utilized chronic ad libitum ethanol (EtOH) feeding (Lieber–DeCarli liquid diet model), chronic intragastric EtOH administration (Tsukamoto–French model), and chronic-plus-binge EtOH challenge (Bin Gao—National Institute on Alcohol Abuse and Alcoholism (NIAAA) model). This review provides an overview of recent advances in rodent models of binge EtOH administration which help to recapitulate different features and etiologies of progressive ALD. These models include EtOH binge alone, and EtOH binge coupled with chronic EtOH intake, a high fat diet, or endotoxin challenge. We analyze the strengths, limitations, and translational relevance of these models, as well as summarize the liver injury outcomes and mechanistic insights. We further discuss the application(s) of binge EtOH models in examining alcohol-induced multi-organ pathology, sex- and age-related differences, as well as circadian rhythm disruption.
Collapse
Affiliation(s)
- Shubha Ghosh Dastidar
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA.
| | - Jeffrey B Warner
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA.
| | - Dennis R Warner
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA.
| | - Craig J McClain
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA.
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA.
- Robley Rex Veterans Medical Center, Louisville, KY 40202, USA.
- University of Louisville Alcohol Research Center and Hepatobiology & Toxicology COBRE, University of Louisville, Louisville, KY 40202, USA.
| | - Irina A Kirpich
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA.
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA.
- University of Louisville Alcohol Research Center and Hepatobiology & Toxicology COBRE, University of Louisville, Louisville, KY 40202, USA.
| |
Collapse
|
21
|
Misale MS, Witek Janusek L, Tell D, Mathews HL. Chromatin organization as an indicator of glucocorticoid induced natural killer cell dysfunction. Brain Behav Immun 2018; 67:279-289. [PMID: 28911980 PMCID: PMC5696065 DOI: 10.1016/j.bbi.2017.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 09/01/2017] [Accepted: 09/08/2017] [Indexed: 11/19/2022] Open
Abstract
It is well-established that psychological distress reduces natural killer cell immune function and that this reduction can be due to the stress-induced release of glucocorticoids. Glucocorticoids are known to alter epigenetic marks associated with immune effector loci, and are also known to influence chromatin organization. The purpose of this investigation was to assess the effect of glucocorticoids on natural killer cell chromatin organization and to determine the relationship of chromatin organization to natural killer cell effector function, e.g. interferon gamma production. Interferon gamma production is the prototypic cytokine produced by natural killer cells and is known to modulate both innate and adaptive immunity. Glucocorticoid treatment of human peripheral blood mononuclear cells resulted in a significant reduction in interferon gamma production. Glucocorticoid treatment also resulted in a demonstrable natural killer cell nuclear phenotype. This phenotype was localization of the histone, post-translational epigenetic mark, H3K27me3, to the nuclear periphery. Peripheral nuclear localization of H3K27me3 was directly related to cellular levels of interferon gamma. This nuclear phenotype was determined by direct visual inspection and by use of an automated, high through-put technology, the Amnis ImageStream. This technology combines the per-cell information content provided by standard microscopy with the statistical significance afforded by large sample sizes common to standard flow cytometry. Most importantly, this technology provides for a direct assessment of the localization of signal intensity within individual cells. The results demonstrate glucocorticoids to dysregulate natural killer cell function at least in part through altered H3K27me3 nuclear organization and demonstrate H3K27me3 chromatin organization to be a predictive indicator of glucocorticoid induced immune dysregulation of natural killer cells.
Collapse
Affiliation(s)
- Michael S Misale
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Health Science Division, 2160 South First Ave., Maywood, IL 60153, United States
| | - Linda Witek Janusek
- Marcella Niehoff School of Nursing, Department of Health Promotion, Loyola University Chicago, Health Science Division, 2160 South First Ave., Maywood, IL 60153, United States
| | - Dina Tell
- Marcella Niehoff School of Nursing, Department of Health Promotion, Loyola University Chicago, Health Science Division, 2160 South First Ave., Maywood, IL 60153, United States
| | - Herbert L Mathews
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Health Science Division, 2160 South First Ave., Maywood, IL 60153, United States.
| |
Collapse
|
22
|
Kalisch-Smith JI, Moritz KM. Detrimental effects of alcohol exposure around conception: putative mechanisms. Biochem Cell Biol 2017; 96:107-116. [PMID: 29112458 DOI: 10.1139/bcb-2017-0133] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In western countries, alcohol consumption is widespread in women of reproductive age, and in binge quantities. These countries also continue to have high incidences of unplanned pregnancies, with women often reported to cease drinking after discovering their pregnancy. This suggests the early embryo may be highly exposed to the detrimental effects of alcohol during the periconception period. The periconception and pre-implantation windows, which include maturation of the oocyte, fertilisation, and morphogenesis of the pre-implantation embryo, are particularly sensitive times of development. Within the oviduct and uterus, the embryo is exposed to a unique nutritional environment to facilitate its development and establish de-novo expression of the genome through epigenetic reprogramming. Alcohol has wide-ranging effects on cellular stress, as well as hormonal, and nutrient signalling pathways, which may affect the development and metabolism of the early embryo. In this review, we summarise the adverse developmental outcomes of early exposure to alcohol (prior to implantation in animal models) and discuss the potential mechanisms for these outcomes that may occur within the protected oviductal and uterine environment. One interesting candidate is reduced retinoic acid synthesis, as it is implicated in the control of epigenetic reprogramming and cell lineage commitment, processes that have adverse consequences for the formation of the placenta, and subsequently, fetal programming.
Collapse
Affiliation(s)
- J I Kalisch-Smith
- a School of Biomedical Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - K M Moritz
- a School of Biomedical Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia.,b Child Health Research Centre, The University of Queensland, South Brisbane, QLD 4101, Australia
| |
Collapse
|
23
|
Banik A, Kandilya D, Ramya S, Stünkel W, Chong YS, Dheen ST. Maternal Factors that Induce Epigenetic Changes Contribute to Neurological Disorders in Offspring. Genes (Basel) 2017; 8:E150. [PMID: 28538662 PMCID: PMC5485514 DOI: 10.3390/genes8060150] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/06/2017] [Accepted: 05/19/2017] [Indexed: 12/12/2022] Open
Abstract
It is well established that the regulation of epigenetic factors, including chromatic reorganization, histone modifications, DNA methylation, and miRNA regulation, is critical for the normal development and functioning of the human brain. There are a number of maternal factors influencing epigenetic pathways such as lifestyle, including diet, alcohol consumption, and smoking, as well as age and infections (viral or bacterial). Genetic and metabolic alterations such as obesity, gestational diabetes mellitus (GDM), and thyroidism alter epigenetic mechanisms, thereby contributing to neurodevelopmental disorders (NDs) such as embryonic neural tube defects (NTDs), autism, Down's syndrome, Rett syndrome, and later onset of neuropsychological deficits. This review comprehensively describes the recent findings in the epigenetic landscape contributing to altered molecular profiles resulting in NDs. Furthermore, we will discuss potential avenues for future research to identify diagnostic markers and therapeutic epi-drugs to reverse these abnormalities in the brain as epigenetic marks are plastic and reversible in nature.
Collapse
Affiliation(s)
- Avijit Banik
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore.
| | - Deepika Kandilya
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore.
| | - Seshadri Ramya
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore.
| | - Walter Stünkel
- Singapore Institute of Clinical Sciences, A*STAR, Singapore 117609, Singapore.
| | - Yap Seng Chong
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore.
| | - S Thameem Dheen
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore.
| |
Collapse
|
24
|
Peana AT, Sánchez-Catalán MJ, Hipólito L, Rosas M, Porru S, Bennardini F, Romualdi P, Caputi FF, Candeletti S, Polache A, Granero L, Acquas E. Mystic Acetaldehyde: The Never-Ending Story on Alcoholism. Front Behav Neurosci 2017; 11:81. [PMID: 28553209 PMCID: PMC5425597 DOI: 10.3389/fnbeh.2017.00081] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 04/18/2017] [Indexed: 12/12/2022] Open
Abstract
After decades of uncertainties and drawbacks, the study on the role and significance of acetaldehyde in the effects of ethanol seemed to have found its main paths. Accordingly, the effects of acetaldehyde, after its systemic or central administration and as obtained following ethanol metabolism, looked as they were extensively characterized. However, almost 5 years after this research appeared at its highest momentum, the investigations on this topic have been revitalized on at least three main directions: (1) the role and the behavioral significance of acetaldehyde in different phases of ethanol self-administration and in voluntary ethanol consumption; (2) the distinction, in the central effects of ethanol, between those arising from its non-metabolized fraction and those attributable to ethanol-derived acetaldehyde; and (3) the role of the acetaldehyde-dopamine condensation product, salsolinol. The present review article aims at presenting and discussing prospectively the most recent data accumulated following these three research pathways on this never-ending story in order to offer the most up-to-date synoptic critical view on such still unresolved and exciting topic.
Collapse
Affiliation(s)
| | - María J. Sánchez-Catalán
- Department of Pharmacy, Pharmaceutical Technology and Parasitology, University of ValenciaValència, Spain
| | - Lucia Hipólito
- Department of Pharmacy, Pharmaceutical Technology and Parasitology, University of ValenciaValència, Spain
| | - Michela Rosas
- Department of Life and Environmental Sciences, University of CagliariCagliari, Italy
| | - Simona Porru
- Department of Life and Environmental Sciences, University of CagliariCagliari, Italy
| | | | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology, University of BolognaBologna, Italy
| | - Francesca F. Caputi
- Department of Pharmacy and Biotechnology, University of BolognaBologna, Italy
| | - Sanzio Candeletti
- Department of Pharmacy and Biotechnology, University of BolognaBologna, Italy
| | - Ana Polache
- Department of Pharmacy, Pharmaceutical Technology and Parasitology, University of ValenciaValència, Spain
| | - Luis Granero
- Department of Pharmacy, Pharmaceutical Technology and Parasitology, University of ValenciaValència, Spain
| | - Elio Acquas
- Department of Life and Environmental Sciences, University of CagliariCagliari, Italy
- Centre of Excellence on Neurobiology of Addiction, University of CagliariCagliari, Italy
| |
Collapse
|
25
|
Chastain LG, Sarkar DK. Alcohol effects on the epigenome in the germline: Role in the inheritance of alcohol-related pathology. Alcohol 2017; 60:53-66. [PMID: 28431793 DOI: 10.1016/j.alcohol.2016.12.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 12/04/2016] [Accepted: 12/30/2016] [Indexed: 12/18/2022]
Abstract
Excessive alcohol exposure has severe health consequences, and clinical and animal studies have demonstrated that disruptions in the epigenome of somatic cells, such as those in brain, are an important factor in the development of alcohol-related pathologies, such as alcohol-use disorders (AUDs) and fetal alcohol spectrum disorders (FASDs). It is also well known that alcohol-related health problems are passed down across generations in human populations, but the complete mechanisms for this phenomenon are currently unknown. Recent studies in animal models have suggested that epigenetic factors are also responsible for the transmission of alcohol-related pathologies across generations. Alcohol exposure has been shown to induce changes in the epigenome of sperm of exposed male animals, and these epimutations are inherited in the offspring. This paper reviews evidence for multigenerational and transgenerational epigenetic inheritance of alcohol-related pathology through the germline. We also review the literature on the epigenetic effects of alcohol exposure on somatic cells in brain, and its contribution to AUDs and FASDs. We note gaps in knowledge in this field, such as the lack of clinical studies in human populations and the lack of data on epigenetic inheritance via the female germline, and we suggest future research directions.
Collapse
Affiliation(s)
- Lucy G Chastain
- The Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, 67 Poultry Lane, New Brunswick, NJ 08901, USA
| | - Dipak K Sarkar
- The Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, 67 Poultry Lane, New Brunswick, NJ 08901, USA.
| |
Collapse
|
26
|
Parira T, Laverde A, Agudelo M. Epigenetic Interactions between Alcohol and Cannabinergic Effects: Focus on Histone Modification and DNA Methylation. JOURNAL OF ALCOHOLISM AND DRUG DEPENDENCE 2017; 5:259. [PMID: 28730160 PMCID: PMC5515243 DOI: 10.4172/2329-6488.1000259] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Epigenetic studies have led to a more profound understanding of the mechanisms involved in chronic conditions. In the case of alcohol addiction, according to the National Institute on Alcohol Abuse and Alcoholism, 16 million adults suffer from Alcohol Use Disorders (AUDs). Even though therapeutic interventions like behavioral therapy and medications to prevent relapse are currently available, no robust cure exists, which stems from the lack of understanding the mechanisms of action of alcohol and the lack of development of precision medicine approaches to treat AUDs. Another common group of addictive substance, cannabinoids, have been studied extensively to reveal they work through cannabinoid receptors. Therapeutic applications have been found for the cannabinoids and a deeper understanding of the endocannabinoid system has been gained over the years. Recent reports of cannabinergic mechanisms in AUDs has opened an exciting realm of research that seeks to elucidate the molecular mechanisms of alcohol-induced end organ diseases and hopefully provide insight into new therapeutic strategies for the treatment of AUDs. To date, several epigenetic mechanisms have been associated with alcohol and cannabinoids independently. Therefore, the scope of this review is to compile the most recent literature regarding alcohol and cannabinoids in terms of a possible epigenetic connection between the endocannabinoid system and alcohol effects. First, we will provide an overview of epigenetics, followed by an overview of alcohol and epigenetic mechanisms with an emphasis on histone modifications and DNA methylations. Then, we will provide an overview of cannabinoids and epigenetic mechanisms. Lastly, we will discuss evidence of interactions between alcohol and cannabinergic pathways and possible insights into the novel epigenetic mechanisms underlying alcohol-cannabinergic pathway activity. Finalizing the review will be a discussion of future directions and therapeutic applications.
Collapse
Affiliation(s)
- Tiyash Parira
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, FL 33199, USA
| | - Alejandra Laverde
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, FL 33199, USA
| | - Marisela Agudelo
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, FL 33199, USA
| |
Collapse
|
27
|
Yan X, Pan B, Lv T, Liu L, Zhu J, Shen W, Huang X, Tian J. Inhibition of histone acetylation by curcumin reduces alcohol-induced fetal cardiac apoptosis. J Biomed Sci 2017; 24:1. [PMID: 28056970 PMCID: PMC5217636 DOI: 10.1186/s12929-016-0310-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 12/05/2016] [Indexed: 02/01/2023] Open
Abstract
Background Prenatal alcohol exposure may cause cardiac development defects, however, the underlying mechanisms are not yet clear. In the present study we have investigated the roles of histone modification by curcumin on alcohol induced fetal cardiac abnormalities during the development. Methods and results Q-PCR and Western blot results showed that alcohol exposure increased gene and active forms of caspase-3 and caspase-8, while decreased gene and protein of bcl-2. ChIP assay results showed that, alcohol exposure increased the acetylation of histone H3K9 near the promoter region of caspase-3 and caspase-8, and decreased the acetylation of histone H3K9 near the promoter region of bcl-2. TUNEL assay data revealed that alcohol exposure increased the apoptosis levels in the embryonic hearts. In vitro experiments demonstrated that curcumin treatment could reverse the up-regulation of active forms of caspase-3 and caspase-8, and down-regulation of bcl-2 induced by alcohol treatment. In addition, curcumin also corrected the high level of histone H3K9 acetylation induced by alcohol. Moreover, the high apoptosis level induced by alcohol was reversed after curcumin treatment in cardiac cells. Conclusions These findings indicate that histone modification may play an important role in mediating alcohol induced fetal cardiac apoptosis, possibly through the up-regulation of H3K9 acetylation near the promoter regions of apoptotic genes. Curcumin treatment may correct alcohol-mediated fetal cardiac apoptosis, suggesting that curcumin may play a protective role against alcohol abuse caused cardiac damage during pregnancy.
Collapse
Affiliation(s)
- Xiaochen Yan
- Department of Cardiology, Heart Centre, The Children's Hospital of Chongqing Medical University, 136 Zhongshan Er Rold, Yu Zhong District, Chongqing, 400014, China.,Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Bo Pan
- Department of Cardiology, Heart Centre, The Children's Hospital of Chongqing Medical University, 136 Zhongshan Er Rold, Yu Zhong District, Chongqing, 400014, China.,Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Tiewei Lv
- Department of Cardiology, Heart Centre, The Children's Hospital of Chongqing Medical University, 136 Zhongshan Er Rold, Yu Zhong District, Chongqing, 400014, China
| | - Lingjuan Liu
- Department of Cardiology, Heart Centre, The Children's Hospital of Chongqing Medical University, 136 Zhongshan Er Rold, Yu Zhong District, Chongqing, 400014, China
| | - Jing Zhu
- Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Wen Shen
- Department of Biomedical Science, Charlie E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Road, Boca Raton, FL, 33431, USA
| | - Xupei Huang
- Department of Biomedical Science, Charlie E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Road, Boca Raton, FL, 33431, USA.
| | - Jie Tian
- Department of Cardiology, Heart Centre, The Children's Hospital of Chongqing Medical University, 136 Zhongshan Er Rold, Yu Zhong District, Chongqing, 400014, China.
| |
Collapse
|
28
|
Shi J, Zhao W, Pan B, Zheng M, Si L, Zhu J, Liu L, Tian J. Alcohol Exposure Causes Overexpression of Heart Development-Related Genes by Affecting the Histone H3 Acetylation via BMP Signaling Pathway in Cardiomyoblast Cells. Alcohol Clin Exp Res 2016; 41:87-95. [PMID: 27883221 DOI: 10.1111/acer.13273] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 10/20/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Abusive alcohol utilization of pregnant woman may cause congenital heart disease (CHD) of fetus, where alcohol ignites histone H3 hyperacetylation leading to abnormal development of heart morphogenesis and associated genes. Knowledge about the regularized upstream genes is little, but bone morphogenetic protein (BMP) signaling may actively and prominently take part in alteration in acetylation of histone H3. The supreme objective of this study was to unearth the involvement of BMP signaling pathway in alcohol-driven hyperacetylation of histone H3 in cardiomyoblast cells. METHODS Cardiomyoblast cells (H9c2 cells) were addicted with alcohol (100 mM) for 24 hours. Dorsomorphin (5 μM) was used for the inhibition of BMP signaling pathway. We detected the phosphorylation activity of SMAD1/5/8, mRNA expression, histone acetyltransferases (HAT)/histone deacetylase (HDAC) activity, and acetylation of histone H3. RESULTS Following alcohol exposure, phosphorylation of SMAD1/5/8 and HAT activities was increased to a significant extent, while histone H3 acetylation and expression of heart development-related genes were also increased. The said phenomenon influenced by alcohol was reverted upon dorsomorphin treatment to the cells without effecting HDAC activity. CONCLUSIONS The data clearly identified that BMP-mediated histone H3 acetylation of heart development-related genes might be one of the possible cellular mechanisms to control alcohol-induced expression of heart development-related genes. Dorsomorphin, on the other hand, may modulate alcohol-induced hyperacetylation of histone H3 through BMP targeting, which could be a potential way to block CHD.
Collapse
Affiliation(s)
- Jin Shi
- Heart Centre, Children's Hospital of Chongqing Medical University, Chongqing, China.,Key Laboratory of Developmental Disease in Childhood, Ministry of Education, Chongqing Medical University, Chongqing, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Weian Zhao
- Heart Centre, Children's Hospital of Chongqing Medical University, Chongqing, China.,Key Laboratory of Developmental Disease in Childhood, Ministry of Education, Chongqing Medical University, Chongqing, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Bo Pan
- Heart Centre, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Min Zheng
- Heart Centre, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Lina Si
- Heart Centre, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Zhu
- Key Laboratory of Developmental Disease in Childhood, Ministry of Education, Chongqing Medical University, Chongqing, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Lingjuan Liu
- Key Laboratory of Developmental Disease in Childhood, Ministry of Education, Chongqing Medical University, Chongqing, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Jie Tian
- Heart Centre, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
29
|
Ethanol downregulates N-acyl phosphatidylethanolamine-phospholipase D expression in BV2 microglial cells via epigenetic mechanisms. Eur J Pharmacol 2016; 786:224-233. [PMID: 27266665 DOI: 10.1016/j.ejphar.2016.06.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 05/30/2016] [Accepted: 06/02/2016] [Indexed: 11/23/2022]
Abstract
Excessive ethanol drinking has deleterious effects on the brain. However, the effects of alcohol on microglia, the main mediator of the brain's innate immune response remain poorly understood. On the other hand, the endocannabinoid system plays a fundamental role in regulating microglial reactivity and function. Here we studied the effects of acute ethanol exposure to murine BV2 microglial cells on N-acyl phosphatidylethanolamine-phospholipase D (NAPE-PLD), a major synthesizing enzyme of anandamide and other N-acylethanolamines. We found that ethanol downregulated microglial NAPE-PLD expression by activating cAMP/PKA and ERK1/2. These signaling pathways converged on increased phosphorylation of CREB. Moreover, ethanol induced and increase in histone acetyltransferase activity which led to higher levels of acetylation of histone H3. Taken together, our results suggest that ethanol actions on microglial NAPE-PLD expression might involve epigenetic mechanisms.
Collapse
|
30
|
Xu YH, Wang LL, Shi L, Lei JP, Miao Q, Liu TQ, Hao W, Lu L, Zhang RL. The association of HTR3A mRNA expression and craving in Han Chinese alcohol-dependent patients: a preliminary study. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2016; 42:316-24. [PMID: 27144979 DOI: 10.3109/00952990.2016.1160098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Ya-Hui Xu
- Department of Addiction, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Lin-Lin Wang
- Department of Psychology, Ninth People Hospital of Zhengzhou, Zhengzhou, Henan Province, China
| | - Le Shi
- Peking University Sixth Hospital/Institute of Mental Health and Key Laboratory of Mental Health and National Institute on Drug Dependence, Peking University, Beijing, China
| | - Jin-Ping Lei
- People Hospital of Cangzhou, Cangzhou, Hebei Province, China
| | - Qin Miao
- Department of Addiction, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Tie-Qiao Liu
- Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Wei Hao
- Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Lin Lu
- Peking University Sixth Hospital/Institute of Mental Health and Key Laboratory of Mental Health and National Institute on Drug Dependence, Peking University, Beijing, China
| | - Rui-Ling Zhang
- Department of Addiction, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province, China
| |
Collapse
|
31
|
Llerena S, Arias-Loste MT, Puente A, Cabezas J, Crespo J, Fábrega E. Binge drinking: Burden of liver disease and beyond. World J Hepatol 2015; 7:2703-2715. [PMID: 26644814 PMCID: PMC4663390 DOI: 10.4254/wjh.v7.i27.2703] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 10/26/2015] [Accepted: 11/11/2015] [Indexed: 02/06/2023] Open
Abstract
The consumption of alcoholic beverages is harmful to human health. In recent years, consumption patterns of alcoholic beverages have changed in our society, and binge drinking has generalized. It is considered to be a socio-sanitary problem with few known consequences in terms of individual and third-party social impacts (in the form of violence or traffic accidents) and its organic impact (affects the liver and other organs and systems, such as the nervous and cardiovascular systems) and represents an important financial burden due to its increasing economic impact. This review provides a global approach to binge drinking and emphasizes its epidemiological character, the effect of this type of consumption and the possible management of a problem with an increasing tendency in our society.
Collapse
|
32
|
In Vivo Acute on Chronic Ethanol Effects in Liver: A Mouse Model Exhibiting Exacerbated Injury, Altered Metabolic and Epigenetic Responses. Biomolecules 2015; 5:3280-94. [PMID: 26610587 PMCID: PMC4693278 DOI: 10.3390/biom5043280] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/17/2015] [Indexed: 01/06/2023] Open
Abstract
Chronic alcoholics who also binge drink (i.e., acute on chronic) are prone to an exacerbated liver injury but its mechanism is not understood. We therefore investigated the in vivo effects of chronic and binge ethanol ingestion and compared to chronic ethanol followed by three repeat binge ethanol on the liver of male C57/BL6 mice fed ethanol in liquid diet (4%) for four weeks followed by binge ethanol (intragastric administration, 3.5 g/kg body weight, three doses, 12h apart). Chronic followed by binge ethanol exacerbated fat accumulation, necrosis, decrease in hepatic SAM and SAM:SAH ratio, increase in adenosine levels, and elevated CYP2E1 levels. Histone H3 lysine acetylation (H3AcK9), dually modified phosphoacetylated histone H3 (H3AcK9/PS10), and phosphorylated H2AX increased after binge whereas phosphorylation of histone H3 ser 10 (H3S10) and H3 ser 28 (H3S28) increased after chronic ethanol-binge. Histone H3 lysine 4 and 9 dimethylation increased with a marked dimethylation in H3K9 in chronic ethanol binge group. Trimethylated histone H3 levels did not change. Nuclear levels of histone acetyl transferase GCN5 and histone deacetylase HDAC3 were elevated whereas phospho-CREB decreased in a distinctive manner. Taken together, acute on chronic ethanol ingestion caused amplification of liver injury and elicited characteristic profiles of histone modifications, metabolic alterations, and changes in nuclear protein levels. These findings demonstrate that chronic ethanol exposure renders liver more susceptible to repeat acute/binge ethanol induced acceleration of alcoholic liver disease.
Collapse
|
33
|
Varadinova M, Boyadjieva N. Epigenetic mechanisms: A possible link between autism spectrum disorders and fetal alcohol spectrum disorders. Pharmacol Res 2015; 102:71-80. [PMID: 26408203 DOI: 10.1016/j.phrs.2015.09.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/12/2015] [Accepted: 09/13/2015] [Indexed: 01/26/2023]
Abstract
The etiology of autism spectrum disorders (ASDs) still remains unclear and seems to involve a considerable overlap between polygenic, epigenetic and environmental factors. We have summarized the current understanding of the interplay between gene expression dysregulation via epigenetic modifications and the potential epigenetic impact of environmental factors in neurodevelopmental deficits. Furthermore, we discuss the scientific controversies of the relationship between prenatal exposure to alcohol and alcohol-induced epigenetic dysregulations, and gene expression alterations which are associated with disrupted neural plasticity and causal pathways for ASDs. The review of the literature suggests that a better understanding of developmental epigenetics should contribute to furthering our comprehension of the etiology and pathogenesis of ASDs and fetal alcohol spectrum disorders.
Collapse
Affiliation(s)
- Miroslava Varadinova
- Department of Pharmacology and Toxicology, Medical Faculty, Medical University, Sofia, Bulgaria.
| | - Nadka Boyadjieva
- Department of Pharmacology and Toxicology, Medical Faculty, Medical University, Sofia, Bulgaria.
| |
Collapse
|
34
|
Drinking beyond a lifetime: New and emerging insights into paternal alcohol exposure on subsequent generations. Alcohol 2015; 49:461-70. [PMID: 25887183 DOI: 10.1016/j.alcohol.2015.02.008] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/10/2015] [Accepted: 02/12/2015] [Indexed: 02/07/2023]
Abstract
Alcohol-use disorder (AUD) is prevalent and associated with substantial socioeconomic costs. While heritability estimates of AUD are ∼50%, identifying specific gene variants associated with risk for AUD has proven challenging despite considerable investment. Emerging research into heritability of complex diseases has implicated transmission of epigenetic variants in the development of behavioral phenotypes, including drug preference and drug-induced behavior. Several recent rodent studies have specifically focused on paternal transmission of epigenetic variants, which is especially relevant because sires are not present for offspring rearing and changes to offspring phenotype are assumed to result from modifications to the sperm epigenome. While considerable interest in paternal transmission of epigenetic variants has emerged recently, paternal alcohol exposures have been studied for 30+ years with interesting behavioral and physiologic effects noted on offspring. However, only recently, with improvements in technology to identify epigenetic modifications in germ cells, has it been possible to identify mechanisms by which paternal ethanol exposure alters offspring behavior. This review presents an overview of epigenetic inheritance in the context of paternal ethanol exposure and suggests future studies to identify specific effects of paternal ethanol exposure on offspring behavior and response to ethanol.
Collapse
|
35
|
Pan B, Zhu J, Lv T, Sun H, Huang X, Tian J. Alcohol consumption during gestation causes histone3 lysine9 hyperacetylation and an alternation of expression of heart development-related genes in mice. Alcohol Clin Exp Res 2015; 38:2396-402. [PMID: 25257289 DOI: 10.1111/acer.12518] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 06/11/2014] [Indexed: 01/19/2023]
Abstract
BACKGROUND Alcohol abuse during gestation may cause congenital heart diseases (CHDs). The underlying mechanisms of alcohol-induced cardiac deformities are still not clear. Recent studies suggest that histone modification may play a crucial role in this pathological process. Moreover, our previous studies reported that ethanol could induce histone3 lysine9 (H3K9) hyperacetylation and overexpression of heart development-related genes in vitro. The aim of this study was to investigate the effect of alcohol consumption during gestation on the imbalance of H3K9 acetylation and the alternation of the expression of heart development-related genes during cardiogenesis. METHODS Pregnant mice were exposed to a single dose of alcohol (10 μl/g/d, 56% alcohol) by gavage every day in the morning from embryo day 7.5 (E7.5) to E15.5. Hematoxylin and eosin (H&E) staining was applied for observing the structure of the embryonic hearts. Western blotting and quantitative real-time polymerase chain reaction were used for detecting the level of H3K9 acetylation and gene expression. Histone acetyltransferase (HAT) and histone deacetylase (HDAC) activities were detected by colorimetric assay and fluorometric assay. RESULTS H&E staining of cardiac tissue showed abnormalities of embryonic hearts at E17.5. The level of H3K9 acetylation reached peak at E17.5 and decreased sharply to a low level at birth and maintained at low level afterward. Alcohol exposure increased H3K9 acetylation at E11.5, E14.5, E17.5, and E18.5, respectively (p < 0.05), and enhanced the expression of Gata4 in the embryonic hearts at E14.5 and E17.5, Mef2c at E14.5, and Nkx2.5 at E14.5 and E17.5, (p < 0.05) but not for Tbx5 (p > 0.05). On embryonic day 17.5, HAT activities of embryonic hearts increased significantly, however alcohol exposure did not alter HDAC activities. CONCLUSIONS These data indicate a time course of H3K9 acetylation change during heart development and demonstrate that alcohol exposure in utero may induce an increase of HAT activities, which results in H3K9 hyperacetylation and an increase of the expression of heart development-related genes. These findings reveal a novel epigenetic mechanism that connects the alcohol consumption during the pregnancy and the development of CHD in the fetus.
Collapse
Affiliation(s)
- Bo Pan
- Key Laboratory of Developmental Disease in Childhood (Chongqing Medical University), Ministry of Education, Chongqing, China; Key Laboratory of Pediatrics in Chongqing, Chongqing, China; Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China; Heart Centre, The Children's Hospital of Chongqing Medical University, Chongqing, China
| | | | | | | | | | | |
Collapse
|
36
|
HDAC7 modulates TNF-α-mediated suppression of Leydig cell steroidogenesis. Mol Cell Biochem 2015; 406:83-90. [DOI: 10.1007/s11010-015-2426-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 04/22/2015] [Indexed: 10/23/2022]
|
37
|
Xu Y, Liu X, Zhang X, Zhang G, Zhang R, Liu T, Hao W. Histone acetylation of the htr3a gene in the prefrontal cortex of Wistar rats regulates ethanol-seeking behavior. Neural Regen Res 2015; 7:1021-8. [PMID: 25722691 PMCID: PMC4341274 DOI: 10.3969/j.issn.1673-5374.2012.13.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 03/24/2012] [Indexed: 11/21/2022] Open
Abstract
Previous reports showed that decreased histone deacetylase activity significantly potentiated the rewarding effects of psychostimulants, and that encoding of the 5-HT3 receptor by the htr3a gene was related to ethanol-seeking behavior. However, the effects of a histone deacetylase inhibitor on ethanol-seeking behavior and epigenetic regulation of htr3a mRNA expression after chronic ethanol exposure are not fully understood. Using quantitative reverse transcription-polymerase chain reaction and chromatin immunoprecipitation analysis, we investigated the effects of chronic ethanol exposure and its interaction with a histone deacetylase inhibitor on histone-acetylation-mediated changes in htr3a mRNA expression in the htr3a promoter region. The conditioned place preference procedure was used to evaluate ethanol-seeking behavior. Chronic exposure to ethanol effectively elicited place conditioning. In the prefrontal cortex, the acetylation of H3K9 and htr3a mRNA expression in the htr3a promoter region were significantly higher in the ethanol group than in the saline group. The histone deacetylase inhibitor sodium butyrate potentiated the effects of ethanol on htr3a mRNA expression and enhanced ethanol-induced conditioned place preferences. These results suggest that ethanol upregulates htr3a levels through mechanisms involving H3K9 acetylation, and that histone acetylation may be a therapeutic target for treating ethanol abuse.
Collapse
Affiliation(s)
- Yahui Xu
- Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Xuebing Liu
- Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Xiaojie Zhang
- Townsend Family Laboratories, Graduate Program in Neuroscience University of British, Columbia 2255 Wesbrook Mall, Vancouver, B.C. V6T 1Z3, Canada
| | - Guanbai Zhang
- Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Ruiling Zhang
- Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| | - Tieqiao Liu
- Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Wei Hao
- Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China ; Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| |
Collapse
|
38
|
Jung SH, Gombojav B, Park EC, Nam CM, Ohrr H, Won JU. Population based study of the association between binge drinking and mortality from cancer of oropharynx and esophagus in Korean men: the Kangwha cohort study. Asian Pac J Cancer Prev 2015; 15:3675-9. [PMID: 24870776 DOI: 10.7314/apjcp.2014.15.8.3675] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
We assessed the association between frequency of heavy binge drinking and mortality from oropharynx and esophagus cancer after controlling for the total volume of alcohol intake among Korean men. The cohort comprised 2,677 male residents in Kangwha County, aged 55 or older in March 1985, for their upper digestive tract cancer mortality for 20.8 years up to December 31, 2005. For daily binge drinkers versus non-drinkers, the hazard ratios (95% Cls) for mortality were 4.82 (1.36, 17.1) and 6.75 (1.45, 31.4) for oropharyngeal and esophageal cancers, respectively. Even after adjusting for the volume of alcohol intake, we found the hazard ratios for frequency of binge drinking and mortality of oropharyngeal or esophageal cancer to not change appreciably: the hazard ratios were 4.90 (1.00, 27.0) and 7.17 (1.02, 50.6), respectively. For esophageal cancer, there was a strong dose-response relationship. The frequency of heavy binge drinking and not just the volume of alcohol intake may increase the risk of mortality from upper digestive tract cancer, particularly esophageal cancer in Korean men. These findings need to be confirmed in further studies with a larger sample size.
Collapse
Affiliation(s)
- Sang Hyuk Jung
- Department of Preventive Medicine, Ewha Womans University, Seoul, Korea E-mail :
| | | | | | | | | | | |
Collapse
|
39
|
Aroor AR, Restrepo RJ, Kharbanda KK, Shukla SD. Epigenetic histone modifications in a clinically relevant rat model of chronic ethanol-binge-mediated liver injury. Hepatol Int 2014. [PMID: 26201320 DOI: 10.1007/s12072-014-9546-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
PURPOSE Ethanol binge augments liver injury after chronic ethanol consumption in humans, but the mechanism behind the enhanced liver injury by ethanol binge is not known. In this study we used a clinically relevant rat model in which liver injury is amplified by binge after chronic ethanol treatment and investigated the importance of histone modifications. METHODS Eight-week-old Sprague-Dawley rats were fed ethanol in a liquid diet for 4 weeks. Control rats were fed an isocaloric liquid diet. This was followed by three binge administrations of ethanol (intragastric 5 g/kg body weight, 12 h apart). In the control, ethanol was replaced by water. Four hours after the last binge administration, liver samples were analyzed for histone modifications and parameters of liver injury. RESULTS Chronic ethanol administration alone caused an increase in histone H3 ser10 and ser28 (H3S10 or S28) phosphorylation, and binge ethanol reduced their levels. Levels of dually modified phosphoacetylated histone H3 (H3AcK9/PS10) increased after acute binge ethanol and remained same after chronic ethanol binge. In contrast, histone H3 lysine-9 acetylation (H3AcK9) was not increased after chronic ethanol but increased significantly after acute binge and chronic ethanol binge. Increase in histone acetylation was accompanied by increased phospho-ERK1/2 in the nuclear extracts. Increased acetylation after chronic ethanol binge was also accompanied by increased protein levels of GCN5 histone acetyl transferase and a modest increase in HDAC3 in the nucleus. Histone lysine-9 dimethylation was significantly increased after chronic ethanol binge. Chronic ethanol binge also resulted in a decrease in the SAM:SAH ratio with a relative decrease of SAM levels and a corresponding increase in SAH levels. CONCLUSIONS Ethanol binge after chronic ethanol altered the profile of site-specific histone modifications and may underlie the mechanism of augmented liver injury by chronic-ethanol-binge-treated rats.
Collapse
Affiliation(s)
- Annayya R Aroor
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Ricardo J Restrepo
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Kusum K Kharbanda
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
| | - Shivendra D Shukla
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, 65212, USA.
| |
Collapse
|
40
|
Kleiber ML, Diehl EJ, Laufer BI, Mantha K, Chokroborty-Hoque A, Alberry B, Singh SM. Long-term genomic and epigenomic dysregulation as a consequence of prenatal alcohol exposure: a model for fetal alcohol spectrum disorders. Front Genet 2014; 5:161. [PMID: 24917881 PMCID: PMC4040446 DOI: 10.3389/fgene.2014.00161] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 05/15/2014] [Indexed: 01/02/2023] Open
Abstract
There is abundant evidence that prenatal alcohol exposure leads to a range of behavioral and cognitive impairments, categorized under the term fetal alcohol spectrum disorders (FASDs). These disorders are pervasive in Western cultures and represent the most common preventable source of neurodevelopmental disabilities. The genetic and epigenetic etiology of these phenotypes, including those factors that may maintain these phenotypes throughout the lifetime of an affected individual, has become a recent topic of investigation. This review integrates recent data that has progressed our understanding FASD as a continuum of molecular events, beginning with cellular stress response and ending with a long-term “footprint” of epigenetic dysregulation across the genome. It reports on data from multiple ethanol-treatment paradigms in mouse models that identify changes in gene expression that occur with respect to neurodevelopmental timing of exposure and ethanol dose. These studies have identified patterns of genomic alteration that are dependent on the biological processes occurring at the time of ethanol exposure. This review also adds to evidence that epigenetic processes such as DNA methylation, histone modifications, and non-coding RNA regulation may underlie long-term changes to gene expression patterns. These may be initiated by ethanol-induced alterations to DNA and histone methylation, particularly in imprinted regions of the genome, affecting transcription which is further fine-tuned by altered microRNA expression. These processes are likely complex, genome-wide, and interrelated. The proposed model suggests a potential for intervention, given that epigenetic changes are malleable and may be altered by postnatal environment. This review accentuates the value of mouse models in deciphering the molecular etiology of FASD, including those processes that may provide a target for the ammelioration of this common yet entirely preventable disorder.
Collapse
Affiliation(s)
- Morgan L Kleiber
- Molecular Genetics Unit, Department of Biology, University of Western Ontario , London, ON, Canada
| | - Eric J Diehl
- Molecular Genetics Unit, Department of Biology, University of Western Ontario , London, ON, Canada
| | - Benjamin I Laufer
- Molecular Genetics Unit, Department of Biology, University of Western Ontario , London, ON, Canada
| | - Katarzyna Mantha
- Molecular Genetics Unit, Department of Biology, University of Western Ontario , London, ON, Canada
| | | | - Bonnie Alberry
- Molecular Genetics Unit, Department of Biology, University of Western Ontario , London, ON, Canada
| | - Shiva M Singh
- Molecular Genetics Unit, Department of Biology, University of Western Ontario , London, ON, Canada
| |
Collapse
|
41
|
Mead EA, Sarkar DK. Fetal alcohol spectrum disorders and their transmission through genetic and epigenetic mechanisms. Front Genet 2014; 5:154. [PMID: 24917878 PMCID: PMC4040491 DOI: 10.3389/fgene.2014.00154] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 05/09/2014] [Indexed: 12/20/2022] Open
Abstract
Fetal alcohol spectrum disorders (FASD) are a group of related conditions that arise from prenatal exposure to maternal consumption of the teratogen, ethanol. It has been estimated that roughly 1% of children in the US suffer from FASD (Sampson etal., 1997), though in some world populations, such as inhabitants of some poorer regions of South Africa, the rate can climb to as high as 20% (May etal., 2013). FASD are the largest cause of mental retardation in U.S. neonates, and ironically, are entirely preventable. FASD have been linked to major changes in the hypothalamic-pituitary-adrenal (HPA) axis, resulting in lifelong impairments through mental disorders, retardation, and sensitivity to stress. FASD are linked to an impaired immune system which consequently leads to an elevated risk of cancer and other diseases. FASD arise from a complex interplay of genetic and epigenetic factors. Here, we review current literature on the topic to tease apart what is known in these areas particularly emphasizing HPA axis dysfunction and how this ties into new studies of transgenerational inheritance in FASD.
Collapse
Affiliation(s)
- Edward A Mead
- Rutgers Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Dipak K Sarkar
- Rutgers Endocrine Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
42
|
Lee DW, Nam YK, Kim TK, Kim JH, Kim SY, Min JW, Lee JH, Kim HY, Kim DJ, Choe BY. Dose-dependent influence of short-term intermittent ethanol intoxication on cerebral neurochemical changes in rats detected by ex vivo proton nuclear magnetic resonance spectroscopy. Neuroscience 2014; 262:107-17. [DOI: 10.1016/j.neuroscience.2013.12.061] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 12/21/2013] [Accepted: 12/27/2013] [Indexed: 01/26/2023]
|
43
|
Krishnan HR, Sakharkar AJ, Teppen TL, Berkel TDM, Pandey SC. The epigenetic landscape of alcoholism. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2014; 115:75-116. [PMID: 25131543 DOI: 10.1016/b978-0-12-801311-3.00003-2] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Alcoholism is a complex psychiatric disorder that has a multifactorial etiology. Epigenetic mechanisms are uniquely capable of accounting for the multifactorial nature of the disease in that they are highly stable and are affected by environmental factors, including alcohol itself. Chromatin remodeling causes changes in gene expression in specific brain regions contributing to the endophenotypes of alcoholism such as tolerance and dependence. The epigenetic mechanisms that regulate changes in gene expression observed in addictive behaviors respond not only to alcohol exposure but also to comorbid psychopathology such as the presence of anxiety and stress. This review summarizes recent developments in epigenetic research that may play a role in alcoholism. We propose that pharmacologically manipulating epigenetic targets, as demonstrated in various preclinical models, hold great therapeutic potential in the treatment and prevention of alcoholism.
Collapse
Affiliation(s)
- Harish R Krishnan
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Amul J Sakharkar
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Tara L Teppen
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Tiffani D M Berkel
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Subhash C Pandey
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA; Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA; Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois, USA.
| |
Collapse
|
44
|
Abstract
The etiology of many brain diseases remains allusive to date after intensive investigation of genomic background and symptomatology from the day of birth. Emerging evidences indicate that a third factor, epigenetics prior to the birth, can exert profound influence on the development and functioning of the brain and over many neurodevelopmental syndromes. This chapter reviews how aversive environmental exposure to parents might predispose or increase vulnerability of offspring to neurodevelopmental deficit through alteration of epigenetics. These epigenetic altering environmental factors will be discussed in the category of addictive agents, nutrition or diet, prescriptive medicine, environmental pollutant, and stress. Epigenetic alterations induced by these aversive environmental factors cover all aspects of epigenetics including DNA methylation, histone modification, noncoding RNA, and chromatin modification. Next, the mechanisms how these environmental inputs influence epigenetics will be discussed. Finally, how environmentally altered epigenetic marks affect neurodevelopment is exemplified by the alcohol-induced fetal alcohol syndrome. It is hoped that a thorough understanding of the nature of prenatal epigenetic inputs will enable researchers with a clear vision to better unravel neurodevelopmental deficit, late-onset neuropsychiatric diseases, or idiosyncratic mental disorders.
Collapse
Affiliation(s)
- Chiao-Ling Lo
- Department of Anatomy and Cell Biology, Stark Neuroscience Research Institute, Indiana Alcohol Research Center, Indiana University School of Medicine, and Department of Psychology, Indiana University Purdue University at Indianapolis, Indianapolis, Indiana, USA
| | - Feng C Zhou
- Department of Anatomy and Cell Biology, Stark Neuroscience Research Institute, Indiana Alcohol Research Center, Indiana University School of Medicine, and Department of Psychology, Indiana University Purdue University at Indianapolis, Indianapolis, Indiana, USA.
| |
Collapse
|
45
|
Neuman MG, French SW, Casey CA, Kharbanda KK, Nanau RM, Rasineni K, McVicker BL, Kong V, Donohue TM. Changes in the pathogenesis of alcohol-induced liver disease — Preclinical studies. Exp Mol Pathol 2013; 95:376-84. [PMID: 24161955 DOI: 10.1016/j.yexmp.2013.10.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Accepted: 10/15/2013] [Indexed: 12/14/2022]
|
46
|
Choi I, Kim P, Joo SH, Kim MK, Park JH, Kim HJ, Ryu JH, Cheong JH, Shin CY. Effects of Preconceptional Ethanol Consumption on ADHD-Like Symptoms in Sprague-Dawley Rat Offsprings. Biomol Ther (Seoul) 2013; 20:226-33. [PMID: 24116300 PMCID: PMC3792223 DOI: 10.4062/biomolther.2012.20.2.226] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 11/30/2011] [Accepted: 12/19/2011] [Indexed: 12/21/2022] Open
Abstract
Ethanol exposure during gestational period is related to growth retardation, morphological abnormality, and even in neurological abnormalities including attention deficit/hyperactivity disorder (ADHD)-like behaviors on offspring. However, relatively little is known about the effects of maternal ethanol consumption prior to conception on their offspring. In this study, we investi-gated whether maternal ethanol administration during preconceptional phase produces ADHD-like behaviors in the rat offspring. Sprague-Dawley (SD) female rats were administrated ethanol via intragastric intubation with dosing regimen of 6 g/kg daily for 10 consecutive days and treated female rats then mated with non-treated male SD rats after 8 weeks. Another group subjected to the same procedure as those conducted on ethanol treated group except the saline administration instead of ethanol. Offspring was tested for their ADHD-like behaviors using open field test, Y maze test and impulsivity test that is performed in the aversive electronic foot shock paradigm. Offspring of preconceptional ethanol treated (EtOH) group showed hyperlocomotive activity, attention deficit and impulsivity. And reduction of striatal dopamine transporter (DAT) level was observed by Western blot in the EtOH group, compared to control (Con) group, while the immunohistochemical analysis exhibited increased expression of norepinephrine transporter (NET) in the frontal cortex. These results suggest that maternal ethanol consumption in the preconceptional phase induces ADHD-like behaviors in offspring that might be related to the abnormal expression of DAT and NET in rat.
Collapse
Affiliation(s)
- Inah Choi
- Center for Neuroscience Research, SMART IBST and Department of Pharmacology, School of Medicine, Konkuk University, Seoul 143-701
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ethanol induces cell cycle arrest and triggers apoptosis via Sp1-dependent p75NTR expression in human neuroblastoma cells. Cell Biol Toxicol 2013; 29:365-80. [PMID: 24026251 DOI: 10.1007/s10565-013-9260-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 08/30/2013] [Indexed: 02/07/2023]
Abstract
Ethanol exposure has deleterious effects on the central nervous system. Although several mechanisms for ethanol-induced damage have been suggested, the precise mechanism underlying ethanol-induced neuronal cell death remains unclear. Recent studies indicate that the p75 neurotrophin receptor (p75NTR) has a critical role in the regulation of neuronal survival. This study was designed to examine the role of p75NTR in ethanol-induced apoptotic signaling in neuroblastoma cells. Ethanol caused highly increased level of p75NTR expression. The use of small interfering RNA to inhibit p75NTR expression markedly attenuated ethanol-induced cell cycle arrest and apoptosis. DNA binding activity of Sp1 was increased by ethanol, whereas inhibition of Sp1 activity by mithramycin, a Sp1 inhibitor, or short hairpin RNA suppressed ethanol-induced p75NTR expression. In addition, inhibitors of casein kinase 2 (CK2) and extracellular signal-regulated kinase (ERK) augmented ethanol-induced p75NTR expression. Our results also demonstrate that inhibition of ERK and CK2 caused a further increase in the activation of the p75NTR proximal promoter induced by ethanol. This increased activation was partially suppressed by the deletion of the Sp1 binding sites. These results suggest that Sp1-mediated p75NTR expression is regulated at least in part by ERK and CK2 pathways. The present study also showed that treatment with ethanol resulted in significant increases in the expression of p21, but not the levels of p53 and p53 target genes such as Bax, Puma, and Bcl-2. Furthermore, the inhibition of p75NTR expression or Sp1 activity suppressed ethanol-induced p21 expression, cell cycle arrest, and apoptosis. These data suggest that ethanol increases p75NTR expression, and CK2 and ERK signaling inversely regulate Sp1-mediated p75NTR expression in ethanol-treated neuroblastoma cells. Thus, our study provides more insight into the mechanisms underlying ethanol actions.
Collapse
|
48
|
Kirpich I, Zhang J, Gobejishvili L, Kharebava G, Barker D, Ghare S, Joshi-Barve S, McClain CJ, Barve S. Binge ethanol-induced HDAC3 down-regulates Cpt1α expression leading to hepatic steatosis and injury. Alcohol Clin Exp Res 2013; 37:1920-9. [PMID: 23905631 DOI: 10.1111/acer.12172] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 04/01/2013] [Indexed: 12/22/2022]
Abstract
BACKGROUND Recently, we have demonstrated that acute alcohol exposure due to binge drinking leads to hepatic steatosis with the deregulation of hepatic histone deacetylase (HDAC) expression. Various class I, II, and IV HDACs were down-regulated, whereas expression of HDAC3 was solely up-regulated. Hence, in the present work, we specifically examined the mechanistic role of HDAC3 in the development of hepatic steatosis occurring in response to binge alcohol administration. METHODS C57BL/6 mice were gavaged 3 times with ethanol (EtOH) at a dose of 4.5 g/kg. HDAC inhibitor, Trichostatin A (TSA) was simultaneously injected intraperitoneally at a dose of 1 mg/kg. Hepatic steatosis, injury, expression of HDAC3 and carnitine palmitoyltransferase 1α (CPT1α) were evaluated. HDAC3 and histone H3 acetylation levels at the Cpt1α promoter were analyzed by chromatin immunoprecipitation (ChIP). RESULTS The binge EtOH-mediated increase in HDAC3 was prevented by simultaneous administration of HDAC inhibitor, TSA, which markedly attenuated hepatic steatosis and injury. Importantly, HDAC3 inhibition was able to normalize the down-regulation of Cpt1α expression. Causal role of HDAC3 in the transcriptional repression of Cpt1α was demonstrated by increased HDAC3 binding at the thyroid receptor element site in the Cpt1α distal promoter region. Further, a resultant decrease in the transcriptionally permissive histone H3 lysine 9 acetylation in the proximal promoter region near the transcriptional start site was observed. Notably, TSA treatment reduced HDAC3 binding and increased H3K9 acetylation at Cpt1α promoter leading to increased Cpt1α expression. These molecular events resulted in attenuation of binge alcohol-induced hepatic steatosis. CONCLUSIONS These findings provide insights into potential epigenetic mechanisms underlying transcriptional regulation of Cpt1α in the hepatic steatosis occurring in response to binge EtOH administration.
Collapse
Affiliation(s)
- Irina Kirpich
- Division of Gastroenterology, Hepatology, and Nutrition , Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky; University of Louisville Alcohol Research Center , Louisville, Kentucky
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Sadrian B, Wilson DA, Saito M. Long-lasting neural circuit dysfunction following developmental ethanol exposure. Brain Sci 2013; 3:704-27. [PMID: 24027632 PMCID: PMC3767176 DOI: 10.3390/brainsci3020704] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 04/10/2013] [Accepted: 04/23/2013] [Indexed: 01/14/2023] Open
Abstract
Fetal Alcohol Spectrum Disorder (FASD) is a general diagnosis for those exhibiting long-lasting neurobehavioral and cognitive deficiencies as a result of fetal alcohol exposure. It is among the most common causes of mental deficits today. Those impacted are left to rely on advances in our understanding of the nature of early alcohol-induced disorders toward human therapies. Research findings over the last decade have developed a model where ethanol-induced neurodegeneration impacts early neural circuit development, thereby perpetuating subsequent integration and plasticity in vulnerable brain regions. Here we review our current knowledge of FASD neuropathology based on discoveries of long-lasting neurophysiological effects of acute developmental ethanol exposure in animal models. We discuss the important balance between synaptic excitation and inhibition in normal neural network function, and relate the significance of that balance to human FASD as well as related disease states. Finally, we postulate that excitation/inhibition imbalance caused by early ethanol-induced neurodegeneration results in perturbed local and regional network signaling and therefore neurobehavioral pathology.
Collapse
Affiliation(s)
- Benjamin Sadrian
- Department of Child and Adolescent Psychiatry, New York University Langone School of Medicine, One Park Avenue, Eighth Floor, New York, NY 10128, USA; E-Mail:
- Nathan Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY 10962, USA; E-Mail:
| | - Donald A. Wilson
- Department of Child and Adolescent Psychiatry, New York University Langone School of Medicine, One Park Avenue, Eighth Floor, New York, NY 10128, USA; E-Mail:
- Nathan Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY 10962, USA; E-Mail:
| | - Mariko Saito
- Nathan Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY 10962, USA; E-Mail:
- Department of Psychiatry, New York University Langone School of Medicine, One Park Avenue, Eighth Floor, New York, NY 10128, USA
| |
Collapse
|
50
|
Abstract
Epigenetic medicine is still in its infancy. To date, only a handful of diseases have documented epigenetic correlates upstream of gene regulation including cancer, developmental syndromes and late-onset diseases. The finding that epigenetic markers are dynamic and heterogeneous at tissue and cellular levels, combined with recent identification of a new form of functionally distinct DNA methylation has opened a wider window for investigators to pry into the epigenetic world. It is anticipated that many diseases will be elucidated through this epigenetic inquiry. In this review, we discuss the normal course of DNA methylation during development, taking alcohol as a demonstrator of the epigenetic impact of environmental factors in disease etiology, particularly the growth retardation and neurodevelopmental deficits of fetal alcohol spectrum disorders.
Collapse
Affiliation(s)
- Marisol Resendiz
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yuanyuan Chen
- Department of Anatomy & Cell Biology, MS508, Indiana University School Medicine, Indianapolis, IN 46202, USA
| | - Nail C Öztürk
- Department of Anatomy & Cell Biology, MS508, Indiana University School Medicine, Indianapolis, IN 46202, USA
- Department of Anatomy, Mersin University School of Medicine, Turkey
| | - Feng C Zhou
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Anatomy & Cell Biology, MS508, Indiana University School Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|